Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Cereb Blood Flow Metab ; 44(7): 1174-1183, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38241458

RESUMO

Intracranial aneurysm rupture causes severe disability and high mortality. Epidemiological studies show a strong association between decreased vitamin D levels and an increase in aneurysm rupture. However, the causality and mechanism remain largely unknown. In this study, we tested whether vitamin D deficiency promotes aneurysm rupture and examined the underlying mechanism for the protective role of vitamin D against the development of aneurysm rupture utilizing a mouse model of intracranial aneurysm. Mice consuming a vitamin D-deficient diet had a higher rupture rate than mice with a regular diet. Vitamin D deficiency increased proinflammatory cytokines in the cerebral arteries. Concurrently, vitamin D receptor knockout mice had a higher rupture rate than the corresponding wild-type littermates. The vitamin D receptors on endothelial and vascular smooth muscle cells, but not on hematopoietic cells, mediated the effect of aneurysm rupture. Our results establish that vitamin D protects against the development of aneurysmal rupture through the vitamin D receptors on vascular endothelial and smooth muscle cells. Vitamin D supplementation may be a viable pharmacologic therapy for preventing aneurysm rupture.


Assuntos
Aneurisma Roto , Aneurisma Intracraniano , Camundongos Knockout , Receptores de Calcitriol , Deficiência de Vitamina D , Vitamina D , Animais , Deficiência de Vitamina D/complicações , Aneurisma Intracraniano/etiologia , Camundongos , Aneurisma Roto/etiologia , Receptores de Calcitriol/metabolismo , Receptores de Calcitriol/genética , Receptores de Calcitriol/deficiência , Vitamina D/uso terapêutico , Vitamina D/sangue , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Citocinas/metabolismo , Camundongos Endogâmicos C57BL , Masculino , Modelos Animais de Doenças , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia
2.
Hypertension ; 81(3): 572-581, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38164754

RESUMO

BACKGROUND: Multiple pathways and factors are involved in the rupture of intracranial aneurysms. The EGFR (epidermal growth factor receptor) has been shown to mediate inflammatory vascular diseases, including atherosclerosis and aortic aneurysm. However, the role of EGFR in mediating intracranial aneurysm rupture and its underlying mechanisms have yet to be determined. Emerging evidence indicates that endoplasmic reticulum (ER) stress might be the link between EGFR activation and the resultant inflammation. ER stress is strongly implicated in inflammation and apoptosis of vascular smooth muscle cells, both of which are key components of the pathophysiology of aneurysm rupture. Therefore, we hypothesized that EGFR activation promotes aneurysmal rupture by inducing ER stress. METHODS: Using a preclinical mouse model of intracranial aneurysm, we examined the potential roles of EGFR and ER stress in developing aneurysmal rupture. RESULTS: Pharmacological inhibition of EGFR markedly decreased the rupture rate of intracranial aneurysms without altering the formation rate. EGFR inhibition also significantly reduced the mRNA (messenger RNA) expression levels of ER-stress markers and inflammatory cytokines in cerebral arteries. Similarly, ER-stress inhibition also significantly decreased the rupture rate. In contrast, ER-stress induction nullified the protective effect of EGFR inhibition on aneurysm rupture. CONCLUSIONS: Our data suggest that EGFR activation is an upstream event that contributes to aneurysm rupture via the induction of ER stress. Pharmacological inhibition of EGFR or downstream ER stress may be a promising therapeutic strategy for preventing aneurysm rupture and subarachnoid hemorrhage.


Assuntos
Aneurisma Roto , Aneurisma Intracraniano , Hemorragia Subaracnóidea , Camundongos , Animais , Aneurisma Intracraniano/prevenção & controle , Aneurisma Intracraniano/genética , Hemorragia Subaracnóidea/prevenção & controle , Aneurisma Roto/metabolismo , Receptores ErbB , RNA Mensageiro , Estresse do Retículo Endoplasmático , Inflamação
3.
Stroke ; 52(8): 2661-2670, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34157864

RESUMO

Background and Purpose: The incidences of intracranial aneurysm and aneurysmal subarachnoid hemorrhage are high in postmenopausal women. Although population-based studies suggest that hormone replacement therapy is beneficial for postmenopausal women with intracranial aneurysms, estrogen replacement may no longer be recommended for the prevention of chronic diseases given its association with adverse outcomes, such as cancer and ischemic stroke. The isoflavone daidzein and its intestinal metabolite equol are bioactive phytoestrogens and potent agonists of estrogen receptors. Given their estrogenic properties, we investigated whether the isoflavones daidzein and equol are protective against the formation and rupture of intracranial aneurysms in a mouse model of the postmenopausal state. Methods: We induced intracranial aneurysms in ovariectomized adult female mice using a combination of induced systemic hypertension and a single injection of elastase into the cerebrospinal fluid. We fed the mice with an isoflavone-free diet with/without daidzein supplementation, or in a combination of intraperitoneal equol, or oral vancomycin treatment. We also used estrogen receptor beta knockout mice. Results: Both dietary daidzein and supplementation with its metabolite, equol, were protective against aneurysm formation in ovariectomized mice. The protective effects of daidzein and equol required estrogen receptor-ß. The disruption of the intestinal microbial conversion of daidzein to equol abolished daidzein's protective effect against aneurysm formation. Mice treated with equol had lower inflammatory cytokines in the cerebral arteries, suggesting that phytoestrogens modulate inflammatory processes important to intracranial aneurysm pathogenesis. Conclusions: Our study establishes that both dietary daidzein and its metabolite, equol, protect against aneurysm formation in ovariectomized female mice through the activation of estrogen receptor-ß and subsequent suppression of inflammation. Dietary daidzein's protective effect required the intestinal conversion to equol. Our results indicate the potential therapeutic value of dietary daidzein and its metabolite, equol, for the prevention of the formation of intracranial aneurysms and related subarachnoid hemorrhage.


Assuntos
Equol/uso terapêutico , Aneurisma Intracraniano/prevenção & controle , Aneurisma Intracraniano/fisiopatologia , Isoflavonas/uso terapêutico , Fitoestrógenos/uso terapêutico , Animais , Equol/farmacologia , Feminino , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/sangue , Isoflavonas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovariectomia/efeitos adversos , Fitoestrógenos/farmacologia
4.
Sci Rep ; 11(1): 7818, 2021 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-33837224

RESUMO

Subarachnoid haemorrhage (SAH) is a type of hemorrhagic stroke that is associated with high morbidity and mortality. New effective treatments are needed to improve outcomes. The pathophysiology of SAH is complex and includes early brain injury and delayed cerebral ischemia, both of which are characterized by blood-brain barrier (BBB) impairment. We isolated brain endothelial cells (BECs) from mice subjected to SAH by injection of blood into the prechiasmatic cistern. We used gene expression profiling to identify 707 unique genes (2.8% of transcripts, 403 upregulated, 304 downregulated, 24,865 interrogated probe sets) that were significantly differentially expressed in mouse BECs after SAH. The pathway involving prostaglandin synthesis and regulation was significantly upregulated after SAH, including increased expression of the Ptgs2 gene and its corresponding COX-2 protein. Celecoxib, a selective COX-2 inhibitor, limited upregulation of Ptgs2 in BECs. In this study, we have defined the gene expression profiling of BECs after experimental SAH and provide further insight into BBB pathophysiology, which may be relevant to other neurological diseases such as traumatic brain injury, brain tumours, ischaemic stroke, multiple sclerosis, and neurodegenerative disorders.


Assuntos
Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Hemorragia Subaracnóidea/genética , Hemorragia Subaracnóidea/metabolismo , Transcriptoma , Animais , Lesões Encefálicas/genética , Lesões Encefálicas/metabolismo , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Celecoxib/uso terapêutico , Ciclo-Oxigenase 2/genética , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Modelos Animais de Doenças , Perfilação da Expressão Gênica/métodos , Camundongos , RNA/genética , RNA/isolamento & purificação , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/metabolismo , Hemorragia Subaracnóidea/tratamento farmacológico , Resultado do Tratamento , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
6.
J Cereb Blood Flow Metab ; 41(8): 1842-1857, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33444089

RESUMO

The distribution and clearance of erythrocytes after subarachnoid hemorrhage (SAH) is poorly understood. We aimed to characterize the distribution of erythrocytes after SAH and the cells involved in their clearance. To visualize erythrocyte distribution, we injected fluorescently-labelled erythrocytes into the prechiasmatic cistern of mice. 10 minutes after injection, we found labelled erythrocytes in the subarachnoid space and ventricular system, and also in the perivascular spaces surrounding large penetrating arterioles. 2 and 5 days after SAH, fluorescence was confined within leptomeningeal and perivascular cells. We identified the perivascular cells as perivascular macrophages based on their morphology, location, Iba-1 immunoreactivity and preferential uptake of FITC-dextran. We subsequently depleted meningeal and perivascular macrophages 2 days before or 3 hours after SAH with clodronate liposomes. At day 5 after SAH, we found increased blood deposition in mice treated prior to SAH, but not those treated after. Treatment post-SAH improved neurological scoring, reduced neuronal cell death and perivascular inflammation, whereas pre-treatment only reduced perivascular inflammation. Our data indicate that after SAH, erythrocytes are distributed throughout the subarachnoid space extending into the perivascular spaces of parenchymal arterioles. Furthermore, meningeal and perivascular macrophages are involved in erythrocyte uptake and play an important role in outcome after SAH.


Assuntos
Macrófagos/fisiologia , Hemorragia Subaracnóidea/patologia , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Eritrócitos/química , Eritrócitos/citologia , Eritrócitos/metabolismo , Gliose , Sistema Glinfático/citologia , Sistema Glinfático/patologia , Macrófagos/citologia , Masculino , Meninges/citologia , Meninges/fisiologia , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Imagem Óptica , Hemorragia Subaracnóidea/metabolismo , Espaço Subaracnóideo/citologia , Espaço Subaracnóideo/patologia
7.
Stroke ; 51(11): 3332-3339, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33019897

RESUMO

BACKGROUND AND PURPOSE: Inflammation has emerged as a key component of the pathophysiology of intracranial aneurysms. Mast cells have been detected in human intracranial aneurysm tissues, and their presence was associated with intramural microhemorrhage and wall degeneration. We hypothesized that mast cells play a critical role in the development of aneurysmal rupture, and that mast cells can be used as a therapeutic target for the prevention of aneurysm rupture. METHODS: Intracranial aneurysms were induced in adult mice using a combination of induced systemic hypertension and a single injection of elastase into the cerebrospinal fluid. Aneurysm formation and rupture were assessed over 3 weeks. Roles of mast cells were assessed using a mast cell stabilizer (cromolyn), a mast cell activator (C48/80), and mice that are genetically lacking mature mast cells (KitW-sh/W-sh mice). RESULTS: Pharmacological stabilization of mast cells with cromolyn markedly decreased the rupture rate of aneurysms (80% versus 19%, n=10 versus n =16) without affecting the aneurysm formation. The activation of mast cells with C48/80 significantly increased the rupture rate of aneurysms (25% versus 100%, n=4 versus n=5) without affecting the overall rate of aneurysm formation. Furthermore, the genetic deficiency of mast cells significantly prevented aneurysm rupture (80% versus 25%, n=10 versus n=8, wild-type versus KitW-sh/W-sh mice). CONCLUSIONS: These results suggest that mast cells play a key role in promoting aneurysm rupture but not formation. Stabilizers of mast cells may have a potential therapeutic value in preventing intracranial aneurysm rupture in patients.


Assuntos
Aneurisma Roto/imunologia , Aneurisma Intracraniano/imunologia , Mastócitos/imunologia , Aneurisma Roto/patologia , Aneurisma Roto/prevenção & controle , Animais , Catepsina G/genética , Quimases/genética , Cromolina Sódica/farmacologia , Modelos Animais de Doenças , Interleucina-6/genética , Aneurisma Intracraniano/patologia , Masculino , Estabilizadores de Mastócitos/farmacologia , Mastócitos/efeitos dos fármacos , Mastócitos/patologia , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Transgênicos , Mutação , Proteínas Proto-Oncogênicas c-kit/genética , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Angiotensina/genética , Hemorragia Subaracnóidea/imunologia , Hemorragia Subaracnóidea/patologia , Hemorragia Subaracnóidea/prevenção & controle , Triptases/genética , Fator de Necrose Tumoral alfa/genética , p-Metoxi-N-metilfenetilamina/farmacologia
8.
J Cereb Blood Flow Metab ; 36(11): 1942-1954, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26661216

RESUMO

Outcome varies among patients with subarachnoid hemorrhage but known prognostic factors explain only a small portion of the variation in outcome. We hypothesized that individual genetic variations influence brain and vascular responses to subarachnoid hemorrhage and investigated this using inbred strains of mice.Subarachnoid hemorrhage was induced in seven inbred and a chromosome 7 substitution strain of mouse. Cerebral blood flow, vasospasm of the middle cerebral artery, and brain injury were assessed. After 48 h of subarachnoid hemorrhage, mice showed significant middle cerebral artery vasospasm that correlated positively with reduction in cerebral blood flow at 45 min. Mice also had increased neuronal injury compared to sham controls; A/J and C57BL/6 J strains represented the most and least severe, respectively. However, brain injury did not correlate with cerebral blood flow reduction at 45 min or with vasospasm at 48 h. Chromosome 7 substitution did not influence the degree of vasospasm or brain injury.Our data suggested that mouse genetic background influences outcome of subarachnoid hemorrhage. Investigations into the genetic factors causing these inter-strain differences may provide insight into the etiology of the brain damage following subarachnoid hemorrhage. These findings also have implications for animal modeling of disease and suggest that genetic differences may also modulate outcome in other cardiovascular diseases.


Assuntos
Circulação Cerebrovascular/genética , Patrimônio Genético , Artéria Cerebral Média/fisiopatologia , Hemorragia Subaracnóidea/genética , Vasoespasmo Intracraniano/genética , Animais , Apoptose/genética , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Especificidade da Espécie , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/patologia , Hemorragia Subaracnóidea/fisiopatologia , Vasoespasmo Intracraniano/etiologia , Vasoespasmo Intracraniano/patologia , Vasoespasmo Intracraniano/fisiopatologia
9.
Stroke ; 46(8): 2260-70, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26138121

RESUMO

BACKGROUND AND PURPOSE: Subarachnoid hemorrhage (SAH) is a complex stroke subtype characterized by an initial brain injury, followed by delayed cerebrovascular constriction and ischemia. Current therapeutic strategies nonselectively curtail exacerbated cerebrovascular constriction, which necessarily disrupts the essential and protective process of cerebral blood flow autoregulation. This study identifies a smooth muscle cell autocrine/paracrine signaling network that augments myogenic tone in a murine model of experimental SAH: it links tumor necrosis factor-α (TNFα), the cystic fibrosis transmembrane conductance regulator, and sphingosine-1-phosphate signaling. METHODS: Mouse olfactory cerebral resistance arteries were isolated, cannulated, and pressurized for in vitro vascular reactivity assessments. Cerebral blood flow was measured by speckle flowmetry and magnetic resonance imaging. Standard Western blot, immunohistochemical techniques, and neurobehavioral assessments were also used. RESULTS: We demonstrate that targeting TNFα and sphingosine-1-phosphate signaling in vivo has potential therapeutic application in SAH. Both interventions (1) eliminate the SAH-induced myogenic tone enhancement, but otherwise leave vascular reactivity intact; (2) ameliorate SAH-induced neuronal degeneration and apoptosis; and (3) improve neurobehavioral performance in mice with SAH. Furthermore, TNFα sequestration with etanercept normalizes cerebral perfusion in SAH. CONCLUSIONS: Vascular smooth muscle cell TNFα and sphingosine-1-phosphate signaling significantly enhance cerebral artery tone in SAH; anti-TNFα and anti-sphingosine-1-phosphate treatment may significantly improve clinical outcome.


Assuntos
Lisofosfolipídeos/biossíntese , Esfingosina/análogos & derivados , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/fisiopatologia , Fator de Necrose Tumoral alfa/biossíntese , Vasoconstrição/fisiologia , Animais , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/fisiologia , Marcação de Genes/métodos , Lisofosfolipídeos/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Técnicas de Cultura de Órgãos , Fenilefrina/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Esfingosina/biossíntese , Esfingosina/deficiência , Hemorragia Subaracnóidea/terapia , Fator de Necrose Tumoral alfa/deficiência , Vasoconstrição/efeitos dos fármacos , Sistema Vasomotor/efeitos dos fármacos , Sistema Vasomotor/fisiologia
10.
Acta Neurochir Suppl ; 120: 29-33, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25366595

RESUMO

Spontaneous subarachnoid hemorrhage (SAH) is a form of hemorrhagic stroke that accounts for approximately 7 % of all strokes worldwide and is associated with mortality in approximately 35 % of cases and morbidity in many of the survivors. Studies have suggested that genetic variations may affect the pathophysiology of SAH. The goal of this study was to investigate the effect of mouse genetic background on brain injury and large artery vasospasm after SAH. SAH was induced in seven inbred strains of mice, and the degree of large artery vasospasm and brain injury was assessed. After 48 h, SAH mice showed a significant reduction in middle cerebral artery diameter and increased neuronal injury in the cerebral cortex compared with sham-operated controls. Mouse strains also demonstrated variable degrees of vasospasm and brain injury. This data suggests that different genetic factors influence how much brain injury and vasospasm occur after SAH. Future investigations may provide insight into the causes of these differences between strains and into which genetic contributors may be responsible for vasospasm and brain injury after SAH.


Assuntos
Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/genética , Vasoespasmo Intracraniano/etiologia , Vasoespasmo Intracraniano/genética , Animais , Lesões Encefálicas/etiologia , Lesões Encefálicas/genética , Modelos Animais de Doenças , Aneurisma Intracraniano/complicações , Aneurisma Intracraniano/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos A , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Degeneração Neural/etiologia , Degeneração Neural/genética , Especificidade da Espécie
11.
Acta Neurochir Suppl ; 120: 81-5, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25366604

RESUMO

INTRODUCTION: Subarachnoid hemorrhage (SAH) can result in significant brain injury. Valproic acid (VPA), a widely-used anti-epileptic drug, was investigated as a possible neuroprotective drug in a prechiasmatic injection model of SAH in mice. METHODS: Mice were randomized to the following experimental groups: SAH, SAH + VPA, Sham, and Sham + VPA. VPA (400 mg/kg) or saline was administered within 30 min of SAH induction and every 12 h thereafter for 48 h. Neurobehavioral assessments using the modified Garcia Score were conducted at 24 and 48 h. Brain injury was assessed at 48 h with fluoro-jade b and caspase-3/NeuN histo- and immunohistochemistry. Vasospasm was assessed in the MCA branches using hematoxylin & eosin histology. RESULTS: SAH mice treated with VPA appeared to have improved neurobehavioral assessments at both 24 and 48 h compared to untreated SAH mice. VPA treatment in SAH mice also significantly decreased the number of degenerating neurons on fluoro-jade b staining. In VPA-treated SAH mice, there was a trend toward a decrease in the number of apoptotic neurons on caspase-3/NeuN immunohistochemistry. VPA did not significantly affect vasospasm. CONCLUSION: This study demonstrated that VPA improves neurological outcome and decreases brain injury in a mouse model of SAH.


Assuntos
Anticonvulsivantes/farmacologia , Fármacos Neuroprotetores/farmacologia , Hemorragia Subaracnóidea/tratamento farmacológico , Ácido Valproico/farmacologia , Animais , Circulação Cerebrovascular/efeitos dos fármacos , Circulação Cerebrovascular/fisiologia , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/patologia , Vasoespasmo Intracraniano/tratamento farmacológico , Vasoespasmo Intracraniano/metabolismo , Vasoespasmo Intracraniano/patologia
12.
J Cereb Blood Flow Metab ; 34(11): 1837-47, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25160671

RESUMO

Brain injury after intracerebral hemorrhage (ICH) occurs in cortex and white matter and may be mediated by blood breakdown products, including hemoglobin and heme. Effects of blood breakdown products, bilirubin and bilirubin oxidation products, have not been widely investigated in adult brain. Here, we first determined the effect of bilirubin and its oxidation products on the structure and function of white matter in vitro using brain slices. Subsequently, we determined whether these compounds have an effect on the structure and function of white matter in vivo. In all, 0.5 mmol/L bilirubin treatment significantly damaged both the function and the structure of myelinated axons but not the unmyelinated axons in brain slices. Toxicity of bilirubin in vitro was prevented by dimethyl sulfoxide. Bilirubin oxidation products (BOXes) may be responsible for the toxicity of bilirubin. In in vivo experiments, unmyelinated axons were found more susceptible to damage from bilirubin injection. These results suggest that unmyelinated axons may have a major role in white-matter damage in vivo. Since bilirubin and BOXes appear in a delayed manner after ICH, preventing their toxic effects may be worth investigating therapeutically. Dimethyl sulfoxide or its structurally related derivatives may have a potential therapeutic value at antagonizing axonal damage after hemorrhagic stroke.


Assuntos
Axônios/metabolismo , Bilirrubina/metabolismo , Lesões Encefálicas/metabolismo , Córtex Cerebral/metabolismo , Hemorragia Cerebral/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Axônios/patologia , Lesões Encefálicas/patologia , Córtex Cerebral/patologia , Hemorragia Cerebral/patologia , Masculino , Camundongos , Oxirredução , Acidente Vascular Cerebral/patologia
13.
J Neurosurg ; 121(6): 1424-31, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24745710

RESUMO

OBJECT: Glutamate is important in the pathogenesis of brain damage after cerebral ischemia and traumatic brain injury. Notably, brain extracellular and cerebrospinal fluid as well as blood glutamate concentrations increase after experimental and clinical trauma. While neurons are one potential source of glutamate, platelets also release glutamate as part of their recruitment and might mediate neuronal damage. This study investigates the hypothesis that platelet microthrombi release glutamate that mediates excitotoxic brain injury and neuron dysfunction after subarachnoid hemorrhage (SAH). METHODS: The authors used two models, primary neuronal cultures exposed to activated platelets, as well as a whole-animal SAH preparation. Propidium iodide was used to evaluate neuronal viability, and surface glutamate receptor staining was used to evaluate the phenotype of platelet-exposed neurons. RESULTS: The authors demonstrate that thrombin-activated platelet-rich plasma releases glutamate, at concentrations that can exceed 300 µM. When applied to neuronal cultures, this activated plasma is neurotoxic, and the toxicity is attenuated in part by glutamate receptor antagonists. The authors also demonstrate that exposure to thrombin-activated platelets induces marked downregulation of the surface glutamate receptor glutamate receptor 2, a marker of excitotoxicity exposure and a possible mechanism of neuronal dysfunction. Linear regression demonstrated that 7 days after SAH in rats there was a strong correlation between proximity to microthrombi and reduction of surface glutamate receptors. CONCLUSIONS: The authors conclude that platelet-mediated microthrombosis contributes to neuronal glutamate receptor dysfunction and might mediate brain injury after SAH.


Assuntos
Plaquetas/fisiologia , Neurônios/fisiologia , Receptores de Glutamato/fisiologia , Hemorragia Subaracnóidea/fisiopatologia , Trombose/fisiopatologia , Animais , Plaquetas/citologia , Comunicação Celular/fisiologia , Ácido Glutâmico/fisiologia , Hemostáticos/farmacologia , Masculino , Microeletrodos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurotoxinas/metabolismo , Técnicas de Cultura de Órgãos , Ativação Plaquetária/fisiologia , Plasma Rico em Plaquetas , Cultura Primária de Células , Ratos Sprague-Dawley , Ratos Wistar , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/patologia , Trombina/farmacologia , Trombose/metabolismo , Trombose/patologia
14.
J Cereb Blood Flow Metab ; 34(1): 108-17, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24064494

RESUMO

Patients with aneurysmal subarachnoid hemorrhage (SAH) frequently have deficits in learning and memory that may or may not be associated with detectable brain lesions. We examined mediators of long-term potentiation after SAH in rats to determine what processes might be involved. There was a reduction in synapses in the dendritic layer of the CA1 region on transmission electron microscopy as well as reduced colocalization of microtubule-associated protein 2 (MAP2) and synaptophysin. Immunohistochemistry showed reduced staining for GluR1 and calmodulin kinase 2 and increased staining for GluR2. Myelin basic protein staining was decreased as well. There was no detectable neuronal injury by Fluoro-Jade B, TUNEL, or activated caspase-3 staining. Vasospasm of the large arteries of the circle of Willis was mild to moderate in severity. Nitric oxide was increased and superoxide anion radical was decreased in hippocampal tissue. Cerebral blood flow, measured by magnetic resonance imaging, and cerebral glucose metabolism, measured by positron emission tomography, were no different in SAH compared with control groups. The results suggest that the etiology of loss of LTP after SAH is not cerebral ischemia but may be mediated by effects of subarachnoid blood such as oxidative stress and inflammation.


Assuntos
Região CA1 Hipocampal/ultraestrutura , Hemorragia Subaracnóidea/patologia , Animais , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/fisiopatologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Modelos Animais de Doenças , Potenciação de Longa Duração/fisiologia , Imageamento por Ressonância Magnética , Masculino , Microscopia Eletrônica de Transmissão , Proteínas Associadas aos Microtúbulos/metabolismo , Proteína Básica da Mielina/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Sprague-Dawley , Receptores de AMPA/metabolismo , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/fisiopatologia
15.
Fluids Barriers CNS ; 10(1): 35, 2013 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-24360130

RESUMO

BACKGROUND: Mouse models are commonly used to study central nervous system disorders, in which cerebrospinal fluid (CSF) drainage may be disturbed. However, mouse CSF drainage into lymphatics has not been thoroughly characterized. We aimed to image this using an in vivo approach that combined quantum dot fluorescent nanoparticles with hyperspectral imaging. FINDINGS: Quantum dot 655 was injected into the CSF of the cisterna magna in seven mice and visualized by in vivo hyperspectral imaging at time points 20 and 40 min, 1, 2, and 6 h after injection. In controls (n = 4), quantum dots were applied directly onto intact dura mater covering the cisterna magna. After imaging, lymph nodes in the neck were harvested and processed post-mortem for histological analysis. After injection into the CSF, quantum dot signal was detected in vivo in submandibular lymph nodes of all mice studied as early as 20 min, but not in controls. Post-mortem gross and histological examination of lymph nodes confirmed in vivo observations. CONCLUSIONS: Non-invasive in vivo hyperspectral imaging is a useful tool to study CSF lymphatic drainage and is relevant to understanding this pathway in CNS disease models.

16.
J Cereb Blood Flow Metab ; 33(7): 1008-14, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23549379

RESUMO

Delayed complications of subarachnoid hemorrhage (SAH) such as angiographic vasospasm, cortical spreading ischemia, microcirculatory dysfunction, and microthrombosis are reported in both patients and animal models of SAH. We demonstrated previously that SAH is associated with increased oxidative stress in the brain parenchyma, and that this correlates with dysfunction of endothelial nitric oxide synthase (eNOS) (homodimeric uncoupling). Uncoupling of eNOS exacerbated oxidative stress and enhanced nitric oxide (NO) depletion, and was associated with multiple secondary complications such as microthrombosis, neuronal apoptosis, and release of reactive oxygen species. Thus, we hypothesized that genetic abbrogation of eNOS would confer a beneficial effect on the brain after SAH. Using a prechiasmatic injection model of SAH, we show here that eNOS knockout (KO) significantly alleviates vasospasm of the middle cerebral artery and reduces superoxide production. Endothelial nitric oxide synthase KO also affected other nitric oxide synthase isoforms. It significantly increases neuron nitric oxide synthase expression but has no effect on inducible nitric oxide synthase. Endothelial nitric oxide synthase KO decreases Zn(2+) release after SAH, reduces microthrombi formation, and prevent neuronal degeneration. This work is consistent with our findings where, after SAH, increased oxidative stress can uncouple eNOS via Zn(2+) thiolate oxidation, or theoretically by depletion or oxidation of tetrahydrobiopterin, resulting in a paradoxical release of superoxide anion radical, further exacerbating oxidative stress and microvascular damage.


Assuntos
Trombose Intracraniana/prevenção & controle , Óxido Nítrico Sintase Tipo III/genética , Hemorragia Subaracnóidea/complicações , Vasoespasmo Intracraniano/prevenção & controle , Animais , Western Blotting , Circulação Cerebrovascular/fisiologia , Modelos Animais de Doenças , Fibrinogênio/metabolismo , Imuno-Histoquímica , Trombose Intracraniana/enzimologia , Trombose Intracraniana/etiologia , Trombose Intracraniana/patologia , Camundongos , Camundongos Knockout , Neurônios/enzimologia , Neurônios/patologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/fisiologia , Estresse Oxidativo/genética , Estresse Oxidativo/fisiologia , Hemorragia Subaracnóidea/enzimologia , Superóxidos/metabolismo , Vasoespasmo Intracraniano/enzimologia , Vasoespasmo Intracraniano/etiologia , Vasoespasmo Intracraniano/patologia , Zinco/metabolismo
17.
Acta Neurochir Suppl ; 115: 185-92, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22890667

RESUMO

Microcirculatory dysfunction may contribute to delayed cerebral ischemia after subarachnoid hemorrhage (SAH). This study investigated structural changes in microvessels and their relationship to brain injury after SAH. We used 15 mice (n = 5 for each group) to create sham, saline-injected (100 µl 0.9% NaCl) or SAH (100 µl autologous blood) model by injection into the prechiasmatic cistern. We sacrificed mice 2 days after surgery and examined the brains using scanning electron microscopy (SEM), transmission electron microscopy (TEM), and immunohistochemical staining of fibrinogen. We assessed neuronal apoptosis by terminal deoxynucleotidyl transferase dUTP (deoxyuridine triphosphate) nick end labeling (TUNEL). Nitric oxide (NO) was measured with 4,5-diaminofluorescein-2-diacetate. TEM and SEM demonstrated that mice with SAH had significantly more of them arterioles with lesion characteristics consistent with microthrombi. Microthrombi number correlated with the number of apoptotic neurons and decreased NO in the brain. In conclusion, SAH causes microthrombosis and constriction of arterioles, which correlates with neuronal death and decreased NO. These data suggest NO depletion may contribute to the formation of microthrombosis and arteriolar constriction, which in turn results in neuronal cell death.


Assuntos
Microvasos/patologia , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/patologia , Trombose/etiologia , Análise de Variância , Animais , Constrição Patológica/fisiopatologia , Modelos Animais de Doenças , Fibrinogênio/metabolismo , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Microcirculação/fisiologia , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Microvasos/metabolismo , Microvasos/ultraestrutura , Neurônios/patologia , Óxido Nítrico/metabolismo , Hemorragia Subaracnóidea/etiologia
18.
Acta Neurochir Suppl ; 115: 247-51, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22890676

RESUMO

Animal models have been developed to simulate angiographic vasospasm secondary to subarachnoid hemorrhage (SAH) and to test pharmacologic treatments. Our aim was to evaluate the effect of pharmacologic treatments that have been tested in humans and in preclinical studies to determine if animal models inform results reported in humans. A systematic review and meta-analysis of SAH studies was performed. We investigated predictors of -translation from animals to humans with multivariate logistic regression. Pharmacologic reduction of vasospasm was effective in mice, rats, rabbits, dogs, nonhuman primates, and humans. Animal studies were generally of poor methodologic quality, and there was evidence of publication bias. Fresh blood injection to simulate SAH (vs. clot placement) and evaluation of vasospasm more than 3 days after SAH were independently associated with successful translation. We conclude that reduction of vasospasm is effective in animals and humans, and that injection of fresh blood and evaluation of vasospasm more than 3 days after SAH may be preferable for preclinical models.


Assuntos
Modelos Animais de Doenças , Hemorragia Subaracnóidea/complicações , Vasodilatadores/uso terapêutico , Vasoespasmo Intracraniano/tratamento farmacológico , Vasoespasmo Intracraniano/etiologia , Animais , Angiografia Cerebral , Humanos , PubMed/estatística & dados numéricos , Vasoconstrição/efeitos dos fármacos , Vasodilatadores/farmacologia
19.
Curr Neurovasc Res ; 9(2): 139-48, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22475399

RESUMO

Nimodipine improved outcome in patients with subarachnoid hemorrhage (SAH) although hypotension limited the dose that could be administered systemically. Subarachnoid delivery of nicardipine or nimodipine may be more efficacious. We tested the efficacy of cisternal application of sustained release nicardipine and nimodipine in SAH in monkeys and dogs, respectively. SAH was created in 13 cynomolgus macaques by placement of autologous blood clot around right middle cerebral, anterior cerebral, and internal carotid arteries. Placebo poly-D,L-lactide coglycolide (PLGA), nicardipine PLGA or mibefradil PLGA was inserted in the clots. Catheter and computed tomography angiography (CTA) were performed at baseline and 7 days later (day 7). Cerebral infarction was assessed on day 7 by magnetic resonance imaging. Six dogs underwent baseline angiography and injection of autologous blood plus PLGA or nimodipine-loaded PLGA microparticles into the cisterna magna. Blood injection was repeated 2 days later and angiography 7 and 14 days later. Animals were euthanized and brains were examined histologically. Cerebrospinal fluid and serum nimodipine concentrations were measured. Nicardipine, but not mibefradil PLGA decreased vasospasm in monkeys (paired t-tests) although there was no significant effect on infarctions see on MRI. In dogs, nimodipine-PLGA produced high local concentrations of nimodipine that were associated with reduced basilar artery vasospasm. No untoward histological effects were observed. There was no reduction in microthrombi in animals treated with nimodipine PLGA compared to placebo PLGA. Site-specific, sustained release formulations of dihydropyridines can deliver high concentrations to the cerebrospinal fluid without causing systemic side effects, and may reduce angiographic vasospasm after SAH. Since nimodipine improves outcome in patients with SAH without necessarily preventing vasospasm, further studies are warranted.


Assuntos
Di-Hidropiridinas/administração & dosagem , Hemorragia Subaracnóidea/fisiopatologia , Vasodilatadores/administração & dosagem , Vasoespasmo Intracraniano/tratamento farmacológico , Vasoespasmo Intracraniano/fisiopatologia , Animais , Preparações de Ação Retardada , Modelos Animais de Doenças , Cães , Feminino , Macaca fascicularis , Nicardipino/administração & dosagem , Nimodipina/administração & dosagem , Placebos , Hemorragia Subaracnóidea/complicações , Vasoespasmo Intracraniano/etiologia
20.
J Cereb Blood Flow Metab ; 32(9): 1645-58, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22534672

RESUMO

Animal models have been developed to simulate angiographic vasospasm secondary to subarachnoid hemorrhage (SAH) and to test pharmacologic treatments. Our aim was to evaluate the effect of pharmacologic treatments that have been tested in humans and in preclinical studies to determine if animal models inform results reported in humans. A systematic review and meta-analysis of SAH studies was performed. We investigated predictors of translation from animals to humans with multivariate logistic regression. Pharmacologic reduction of vasospasm was effective in mice, rats, rabbits, dogs, nonhuman primates (standard mean difference of -1.74; 95% confidence interval -2.04 to -1.44) and humans. Animal studies were generally of poor methodologic quality and there was evidence of publication bias. Subgroup analysis by drug and species showed that statins, tissue plasminogen activator, erythropoietin, endothelin receptor antagonists, calcium channel antagonists, fasudil, and tirilazad were effective whereas magnesium was not. Only evaluation of vasospasm >3 days after SAH was independently associated with successful translation. We conclude that reduction of vasospasm is effective in animals and humans and that evaluation of vasospasm >3 days after SAH may be preferable for preclinical models.


Assuntos
Angiografia Cerebral , Hemorragia Subaracnóidea/tratamento farmacológico , Vasoespasmo Intracraniano/tratamento farmacológico , Animais , Interpretação Estatística de Dados , Modelos Animais de Doenças , Cães , Feminino , Humanos , Macaca , Masculino , Camundongos , Viés de Publicação , Coelhos , Ensaios Clínicos Controlados Aleatórios como Assunto , Ratos , Especificidade da Espécie , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/patologia , Resultado do Tratamento , Vasoespasmo Intracraniano/etiologia , Vasoespasmo Intracraniano/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA